1
|
Aboy-Pardal MCM, Guadamillas MC, Guerrero CR, Català-Montoro M, Toledano-Donado M, Terrés-Domínguez S, Pavón DM, Jiménez-Jiménez V, Jimenez-Carretero D, Zamai M, Folgueira C, Cerezo A, Lolo FN, Nogueiras R, Sabio G, Sánchez-Álvarez M, Echarri A, Garcia R, Del Pozo MA. Plasma membrane remodeling determines adipocyte expansion and mechanical adaptability. Nat Commun 2024; 15:10102. [PMID: 39609408 PMCID: PMC11605069 DOI: 10.1038/s41467-024-54224-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Adipocytes expand massively to accommodate excess energy stores and protect the organism from lipotoxicity. Adipose tissue expandability is at the center of disorders such as obesity and lipodystrophy; however, little is known about the relevance of adipocyte biomechanics on the etiology of these conditions. Here, we show in male mice in vivo that the adipocyte plasma membrane undergoes caveolar domain reorganization upon lipid droplet expansion. As the lipid droplet grows, caveolae disassemble to release their membrane reservoir and increase cell surface area, and transfer specific caveolar components to the LD surface. Adipose tissue null for caveolae is stiffer, shows compromised deformability, and is prone to rupture under mechanical compression. Mechanistically, phosphoacceptor Cav1 Tyr14 is required for caveolae disassembly: adipocytes bearing a Tyr14Phe mutation at this residue are stiffer and smaller, leading to decreased adiposity in vivo; exhibit deficient transfer of Cav1 and EHD2 to the LD surface, and show distinct Cav1 molecular dynamics and tension adaptation. These results indicate that Cav1 phosphoregulation modulates caveolar dynamics as a relevant component of the homeostatic mechanoadaptation of the differentiated adipocyte.
Collapse
Affiliation(s)
- María C M Aboy-Pardal
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta C Guadamillas
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Science and Agroforestal Technology and Genetics, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain
| | - Carlos R Guerrero
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Mauro Català-Montoro
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mónica Toledano-Donado
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sara Terrés-Domínguez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Dácil M Pavón
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Allergy Therapeutics, Avenida Punto Es, 12, 28805 Alcalá de Henares, Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Health Science, Universidad Católica Santa Teresa de Jesús de Ávila, Ávila, Spain
| | - Daniel Jimenez-Carretero
- Cellomics Unit, Cell and Developmental Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Moreno Zamai
- Microscopy and Dynamic Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cintia Folgueira
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Cerezo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Molecular Pharmacology, Lilly Research Laboratories, Alcobendas, Spain
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rubén Nogueiras
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
| | - Guadalupe Sabio
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Cell Compartmentalization, Homeostasis and Inflammation lab, Department of Metabolic and Inflammatory Diseases. Instituto de Investigaciones Biomédicas "Sols-Morreale"-CSIC, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Mechanobiology of Organelles lab. Department of Cellular and Molecular Biology. Centro de Investigaciones Biológicas Margarita Salas - CSIC, Madrid, Spain
| | - Ricardo Garcia
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
2
|
Mironov AA, Beznoussenko GV. Algorithm for Modern Electron Microscopic Examination of the Golgi Complex. Methods Mol Biol 2022; 2557:161-209. [PMID: 36512216 DOI: 10.1007/978-1-0716-2639-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Golgi complex (GC) is an essential organelle of the eukaryotic exocytic pathway. It has a very complexed structure and thus localization of its resident proteins is not trivial. Fast development of microscopic methods generates a huge difficulty for Golgi researchers to select the best protocol to use. Modern methods of light microscopy, such as super-resolution light microscopy (SRLM) and electron microscopy (EM), open new possibilities in analysis of various biological structures at organelle, cell, and organ levels. Nowadays, new generation of EM methods became available for the study of the GC; these include three-dimensional EM (3DEM), correlative light-EM (CLEM), immune EM, and new estimators within stereology that allow realization of maximal goal of any morphological study, namely, to achieve a three-dimensional model of the sample with optimal level of resolution and quantitative determination of its chemical composition. Methods of 3DEM have partially overlapping capabilities. This requires a careful comparison of these methods, identification of their strengths and weaknesses, and formulation of recommendations for their application to cell or tissue samples. Here, we present an overview of 3DEM methods for the study of the GC and some basics for how the images are formed and how the image quality can be improved.
Collapse
|
3
|
Aboy-Pardal MC, Jimenez-Carretero D, Terrés-Domínguez S, Pavón DM, Sotodosos-Alonso L, Jiménez-Jiménez V, Sánchez-Cabo F, Del Pozo MA. A deep learning-based tool for the automated detection and analysis of caveolae in transmission electron microscopy images. Comput Struct Biotechnol J 2022; 21:224-237. [PMID: 36544477 PMCID: PMC9755247 DOI: 10.1016/j.csbj.2022.11.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Caveolae are nanoscopic and mechanosensitive invaginations of the plasma membrane, essential for adipocyte biology. Transmission electron microscopy (TEM) offers the highest resolution for caveolae visualization, but provides complicated images that are difficult to classify or segment using traditional automated algorithms such as threshold-based methods. As a result, the time-consuming tasks of localization and quantification of caveolae are currently performed manually. We used the Keras library in R to train a convolutional neural network with a total of 36,000 TEM image crops obtained from adipocytes previously annotated manually by an expert. The resulting model can differentiate caveolae from non-caveolae regions with a 97.44% accuracy. The predictions of this model are further processed to obtain caveolae central coordinate detection and cytoplasm boundary delimitation. The model correctly finds negligible caveolae predictions in images from caveolae depleted Cav1-/- adipocytes. In large reconstructions of adipocyte sections, model and human performances are comparable. We thus provide a new tool for accurate caveolae automated analysis that could speed up and assist in the characterization of the cellular mechanical response.
Collapse
Affiliation(s)
- María C.M. Aboy-Pardal
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| | - Daniel Jimenez-Carretero
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares
(CNIC), 28029 Madrid, Spain
| | - Sara Terrés-Domínguez
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| | - Dácil M. Pavón
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| | - Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares
(CNIC), 28029 Madrid, Spain
| | - Miguel A. Del Pozo
- Mechanoadaptation and Caveolae Biology lab, Cell and Developmental
Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029
Madrid, Spain
| |
Collapse
|
4
|
Ghinea N. Anti-Angiogenic Therapy: Albumin-Binding Proteins Could Mediate Mechanisms Underlying the Accumulation of Small Molecule Receptor Tyrosine Kinase Inhibitors in Normal Tissues with Potential Harmful Effects on Health. Diseases 2021; 9:28. [PMID: 33920299 PMCID: PMC8167546 DOI: 10.3390/diseases9020028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Anti-angiogenics currently used in cancer therapy target angiogenesis by two major mechanisms: (i) neutralizing angiogenic factors or their receptors by using macromolecule anti-angiogenic drugs (e.g., therapeutic antibodies), and (ii) blocking intracellularly the activity of receptor tyrosine kinases with small molecule (Mr < 1 kDa) inhibitors. Anti-angiogenics halt the growth and spread of cancer, and significantly prolong the disease-free survival of the patients. However, resistance to treatment, insufficient efficacy, and toxicity limit the success of this antivascular therapy. Published evidence suggests that four albumin-binding proteins (ABPs) (gp18, gp30, gp60/albondin, and secreted protein acidic and cysteine-rich (SPARC)) could be responsible for the accumulation of small molecule receptor tyrosine kinase inhibitors (RTKIs) in normal organs and tissues and therefore responsible for the side effects and toxicity associated with this type of cancer therapy. Drawing attention to these studies, this review discusses the possible negative role of albumin as a drug carrier and the rationale for a new strategy for cancer therapy based on follicle-stimulating hormone receptor (FSHR) expressed on the luminal endothelial cell surface of peritumoral blood vessels associated with the major human cancers. This review should be relevant to the audience and the field of cancer therapeutics and angiogenesis/microvascular modulation-based interventions.
Collapse
Affiliation(s)
- Nicolae Ghinea
- Research Center, Translational Research Department, Curie Institute, Tumor Angiogenesis Team, 75005 Paris, France
| |
Collapse
|
5
|
|
6
|
Kim SM, Faix PH, Schnitzer JE. Overcoming key biological barriers to cancer drug delivery and efficacy. J Control Release 2017; 267:15-30. [PMID: 28917530 PMCID: PMC8756776 DOI: 10.1016/j.jconrel.2017.09.016] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 01/08/2023]
Abstract
Poor delivery efficiency continues to hamper the effectiveness of cancer therapeutics engineered to destroy solid tumors using different strategies such as nanocarriers, targeting agents, and matching treatments to specific genetic mutations. All contemporary systemic anti-cancer agents are dependent upon passive transvascular mechanisms for their delivery into solid tumors. The therapeutic efficacies of our current drug arsenal could be significantly improved with an active delivery strategy. Here, we discuss how drug delivery and therapeutic efficacy are greatly hindered by barriers presented by the vascular endothelial cell layer and by the aberrant nature of tumor blood vessels in general. We describe mechanisms by which molecules cross endothelial cell (EC) barriers in normal tissues and in solid tumors, including paracellular and transcellular pathways that enable passive or active transport. We also discuss specific obstacles to drug delivery that make solid tumors difficult to treat, as well strategies to overcome them and enhance drug penetration. Finally, we describe the caveolae pumping system, a promising active transport alternative to passive drug delivery across the endothelial cell barrier. Each strategy requires further testing to define its therapeutic applicability and clinical utilities.
Collapse
Affiliation(s)
- Susy M Kim
- Proteogenomics Research Institute for Systems Medicine, 505 Coast Blvd. South, La Jolla, CA 92037, United States
| | - Peggy H Faix
- Proteogenomics Research Institute for Systems Medicine, 505 Coast Blvd. South, La Jolla, CA 92037, United States
| | - Jan E Schnitzer
- Proteogenomics Research Institute for Systems Medicine, 505 Coast Blvd. South, La Jolla, CA 92037, United States.
| |
Collapse
|
7
|
Fullstone G, Nyberg S, Tian X, Battaglia G. From the Blood to the Central Nervous System: A Nanoparticle's Journey Through the Blood-Brain Barrier by Transcytosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 130:41-72. [PMID: 27678174 DOI: 10.1016/bs.irn.2016.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Designing nanoparticles that effectively enter the central nervous system (CNS) rapidly and without alteration is one of the major challenges in the use of nanotechnology for the brain. In this chapter, we explore the process of transcytosis, a receptor-mediated transport pathway that permits endogenous macromolecules to enter the CNS by crossing the blood-brain barrier. Transcytosis across the blood-brain barrier involves a number of distinct stages, including receptor binding, endocytosis into a transport vesicle, trafficking of the vesicle to the opposite side of the cell, and finally exocytosis and release of cargo. For each stage, we discuss the current knowledge on biological, physiological, and physical factors that influence nanoparticle transit through that stage of transcytosis, with implications for nanoparticle design. Finally, we look at the current progress in designing nanoparticles that exploit transcytosis for CNS delivery.
Collapse
Affiliation(s)
- G Fullstone
- University College London, London, United Kingdom.
| | - S Nyberg
- University College London, London, United Kingdom; Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.
| | - X Tian
- School of Life Sciences, Anhui University, Hefei, People's Republic of China
| | - G Battaglia
- University College London, London, United Kingdom.
| |
Collapse
|
8
|
Klaassen I, Van Noorden CJF, Schlingemann RO. Molecular basis of the inner blood-retinal barrier and its breakdown in diabetic macular edema and other pathological conditions. Prog Retin Eye Res 2013; 34:19-48. [PMID: 23416119 DOI: 10.1016/j.preteyeres.2013.02.001] [Citation(s) in RCA: 502] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/19/2012] [Accepted: 02/01/2013] [Indexed: 12/16/2022]
Abstract
Breakdown of the inner endothelial blood-retinal barrier (BRB), as occurs in diabetic retinopathy, age-related macular degeneration, retinal vein occlusions, uveitis and other chronic retinal diseases, results in vasogenic edema and neural tissue damage, causing loss of vision. The central mechanism of altered BRB function is a change in the permeability characteristics of retinal endothelial cells caused by elevated levels of growth factors, cytokines, advanced glycation end products, inflammation, hyperglycemia and loss of pericytes. Subsequently, paracellular but also transcellular transport across the retinal vascular wall increases via opening of endothelial intercellular junctions and qualitative and quantitative changes in endothelial caveolar transcellular transport, respectively. Functional changes in pericytes and astrocytes, as well as structural changes in the composition of the endothelial glycocalyx and the basal lamina around BRB endothelium further facilitate BRB leakage. As Starling's rules apply, active transcellular transport of plasma proteins by the BRB endothelial cells causing increased interstitial osmotic pressure is probably the main factor in the formation of macular edema. The understanding of the complex cellular and molecular processes involved in BRB leakage has grown rapidly in recent years. Although appropriate animal models for human conditions like diabetic macular edema are lacking, these insights have provided tools for rational design of drugs aimed at restoring the BRB as well as for design of effective transport of drugs across the BRB, to treat the chronic retinal diseases such as diabetic macular edema that affect the quality-of-life of millions of patients.
Collapse
Affiliation(s)
- Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
9
|
Abstract
In this issue of Microcirculation, Wagner, Modla, Hossler and Czmmek [25] describe the use of electron tomography to visualize the three-dimensional arrangement of small endothelial vesicles and caveolae of muscle capillaries. Their images show the well-known clusters of fused vesicles communicating with caveolae at the luminal and abluminal surfaces. The advantages of electron tomography are shown by well resolved images of single cytoplasmic vesicles separate from fused vesicle clusters and also by occasional chains of fused vesicles forming trans-endothelial channels. Twenty five to thirty years ago the existence of both trans-endothelial channels and single unattached vesicles was disputed. Also, since some single vesicles and all of the trans-endothelial channels are labeled with a lanthanide tracer present in the perfusate at the time of fixation, this evidence once again raises the question of whether vesicles have a role in vascular permeability to macromolecules. This brief review describes the origin of the vesicle controversy, some of the more recent evidence for and against the participation of vesicles in macromolecular transport and considers some criticisms of ultra-structural evidence for vesicular transport that still require answers.
Collapse
|
10
|
Wagner R, Modla S, Hossler F, Czymmek K. Three-dimensional analysis and computer modeling of the capillary endothelial vesicular system with electron tomography. Microcirculation 2012; 19:477-84. [PMID: 22435898 DOI: 10.1111/j.1549-8719.2012.00181.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE We examined the three-dimensional organization of the endothelial vesicular system with TEM tomography of semi-thick sections. MATERIALS AND METHODS Mouse abdominal muscle capillaries were perfused with terbium to label vesicular compartments open to the luminal surface. The tissue was prepared for TEM and semi-thick (250 nm) sections were cut. Dual axis tilt series, collected from +60° to -60° at 1° increments, were acquired in regions of labeled abluminal caveolae. These tomograms were reconstructed and analyzed to reveal three-dimensional vesicular associations not evident in thin sections. RESULTS Reconstructed tomograms revealed free vesicles, both labeled and unlabeled, in the endothelial cytoplasm as well as transendothelial channels that spanned the luminal and abluminal membranes. A large membranous compartment connecting the luminal and abluminal surfaces was also present. Computer modeling of tomographic data and video animations provided three-dimensional perspectives to these structures. CONCLUSIONS Uncertainties associated with other three-dimensional methods to study the capillary wall are remedied by tomographic analysis of semi-thick sections. Transendothelial channels of fused vesicles and free cytoplasmic vesicles give credence to their role as large pores in the transport of solutes across the walls of continuous capillaries.
Collapse
Affiliation(s)
- Roger Wagner
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, USA.
| | | | | | | |
Collapse
|
11
|
|
12
|
Stender S. The in vivo transfer of free and esterified cholesterol from plasma into the arterial wall of hypercholesterolemic rabbits. Scandinavian Journal of Clinical and Laboratory Investigation 2010. [DOI: 10.1080/00365518209168399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
13
|
Abstract
We have recently shown that an entire oxytocin (OT) system, a peptide and its cognate receptors, is synthesized in the heart. In fetal and newborn hearts, OT exists in its extended three-amino acid form, OT-Gly-Lys-Arg (OT-GKR). OT translocates glucose transporter type 4 to the plasma membrane in human endothelial cells. Therefore, we hypothesized that the cardiac OT/OT-GKR system may be involved in the regulation of myocardial glucose uptake in physiological conditions and during metabolic stress such as hypoxia. Primary cultures of neonatal rat cardiomyocytes (CM) and cardiac progenitor cells expressing ATP-binding cassette efflux transporter G2 transporter (stem cell marker) were studied. OT (10 nm) increased basal glucose uptake in CM to 4.0 +/- 0.2 fmol/mg protein, with OT-GKR (10 nm) elevating it to 5.3 +/- 0.4 fmol/mg protein (P < 0.001) in comparison with 2.2 fmol/mg in control cells. OT had a moderate synergistic effect with 0.1 mm 2,4-dinitrophenol, augmenting basal glucose uptake to 5.5 +/- 0.5 fmol/mg. OT-GKR (10 nm) was even more potent in combination with 2,4-dinitrophenol, increasing glucose uptake to 9.0 +/- 1.0 fmol/mg. Wortmannin (0.1 microm), an inhibitor of phosphatidylinositol-3-kinase, significantly suppressed the effect of OT and insulin (10 nm) (P < 0.001), indicating common pathways. Our data suggest that OT and OT-GKR influence glucose uptake in neonatal rat CM and may thus play a role in the maintenance of cardiac function and cell survival during metabolic stress.
Collapse
Affiliation(s)
- Maria Florian
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Hôtel-Dieu, Pavillon Masson, 3850 Saint-Urbain Street, Montreal, Quebec, Canada
| | | | | |
Collapse
|
14
|
Hansson GK, Bondjers G. Endothelial dysfunction and injury in atherosclerosis. ACTA MEDICA SCANDINAVICA. SUPPLEMENTUM 2009; 715:11-7. [PMID: 2954434 DOI: 10.1111/j.0954-6820.1987.tb09897.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
15
|
|
16
|
|
17
|
Oh P, Borgström P, Witkiewicz H, Li Y, Borgström BJ, Chrastina A, Iwata K, Zinn KR, Baldwin R, Testa JE, Schnitzer JE. Live dynamic imaging of caveolae pumping targeted antibody rapidly and specifically across endothelium in the lung. Nat Biotechnol 2007; 25:327-37. [PMID: 17334358 PMCID: PMC1979160 DOI: 10.1038/nbt1292] [Citation(s) in RCA: 231] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2006] [Accepted: 02/01/2007] [Indexed: 11/09/2022]
Abstract
How effectively and quickly endothelial caveolae can transcytose in vivo is unknown, yet critical for understanding their function and potential clinical utility. Here we use quantitative proteomics to identify aminopeptidase P (APP) concentrated in caveolae of lung endothelium. Electron microscopy confirms this and shows that APP antibody targets nanoparticles to caveolae. Dynamic intravital fluorescence microscopy reveals that targeted caveolae operate effectively as pumps, moving antibody within seconds from blood across endothelium into lung tissue, even against a concentration gradient. This active transcytosis requires normal caveolin-1 expression. Whole body gamma-scintigraphic imaging shows rapid, specific delivery into lung well beyond that achieved by standard vascular targeting. This caveolar trafficking in vivo may underscore a key physiological mechanism for selective transvascular exchange and may provide an enhanced delivery system for imaging agents, drugs, gene-therapy vectors and nanomedicines. 'In vivo proteomic imaging' as described here integrates organellar proteomics with multiple imaging techniques to identify an accessible target space that includes the transvascular pumping space of the caveola.
Collapse
Affiliation(s)
- Phil Oh
- Sidney Kimmel Cancer Center, 10905 Road to the Cure, San Diego, California 92121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pelkmans L. Secrets of caveolae- and lipid raft-mediated endocytosis revealed by mammalian viruses. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:295-304. [PMID: 16126288 DOI: 10.1016/j.bbamcr.2005.06.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Revised: 06/13/2005] [Accepted: 06/15/2005] [Indexed: 11/30/2022]
Abstract
In recent years, it has been unambiguously shown that caveolae and lipid rafts can internalize cargo upon stimulation by multivalent ligands, demonstrated by the infectious entry routes of certain non-enveloped viruses that bind integrins or glycosphingolipids. We currently understand little about the membrane trafficking principles of this endocytic route, but it is clear that we cannot use paradigms from classical membrane traffic. Recent evidence indicates that caveolae- and lipid raft-mediated endocytosis plays important roles in cell adhesion and anchorage-dependent cell growth, but the underlying mechanisms are not known. In this review, I will introduce new models based on current research that aims at identifying the core machinery, regulatory components and design principles of this endocytic route in order to understand its role in cellular physiology. Again, viruses are proving to be excellent tools to reach that goal.
Collapse
Affiliation(s)
- Lucas Pelkmans
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
19
|
Stan RV. Structure of caveolae. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:334-48. [PMID: 16214243 DOI: 10.1016/j.bbamcr.2005.08.008] [Citation(s) in RCA: 210] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2005] [Revised: 08/25/2005] [Accepted: 08/27/2005] [Indexed: 12/11/2022]
Abstract
The introduction of the electron microscope to the study of the biological materials in the second half of the last century has dramatically expanded our view and understanding of the inner workings of cells by enabling the discovery and study of subcellular organelles. A population of flask-shaped or spherical invaginations of the plasma membrane were described and named plasmalemmal vesicles or caveolae. Until the discovery of caveolin-1 as their first molecular marker in early 1990s, the study of caveolae was the exclusive domain of electron microscopists that demonstrated caveolae at different surface densities in most mammalian cells with few exceptions. Electron microscopy techniques in combination with other approaches have also revealed the structural features of caveolae as well as some of their protein and lipid residents. This review summarizes the data on the structure and components of caveolae and their stomatal diaphragms.
Collapse
Affiliation(s)
- Radu V Stan
- Angiogenesis Research Center, Department of Pathology, Dartmouth Medical School, One Medical Center Drive, Lebanon, NH 03756, USA.
| |
Collapse
|
20
|
Predescu D, Vogel SM, Malik AB. Functional and morphological studies of protein transcytosis in continuous endothelia. Am J Physiol Lung Cell Mol Physiol 2004; 287:L895-901. [PMID: 15475492 DOI: 10.1152/ajplung.00075.2004] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Continuous microvascular endothelium constitutively transfers protein from vessel lumen to interstitial space. Compelling recent biochemical, ultrastructural, and physiological evidence reviewed herein demonstrates that protein transport is not the result of barrier “leakiness” but, rather, is an active process occurring primarily in a transendothelial vesicular pathway. Protein accesses the vesicular pathway by means of caveolae open to the vessel lumen. Vascular tracer proteins appear in free cytoplasmic vesicles within minutes; contents of transport vesicles are rapidly deposited into the subendothelial matrix by exocytosis. Caveolin-1 deficiency eliminates caveolae and abolishes vesicular protein transport; interestingly, exchange vessels develop a compensatory transport mode through the opening of a paracellular permeability pathway. The evidence supports the transcytosis hypothesis and the concept that transcytosis is a fundamental component of transendothelial permeability of macromolecules.
Collapse
Affiliation(s)
- Dan Predescu
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | | | | |
Collapse
|
21
|
Guthmann F, Schachtrup C, Tölle A, Wissel H, Binas B, Kondo H, Owada Y, Spener F, Rüstow B. Phenotype of palmitic acid transport and of signalling in alveolar type II cells from E/H-FABP double-knockout mice: contribution of caveolin-1 and PPARgamma. Biochim Biophys Acta Mol Cell Biol Lipids 2004; 1636:196-204. [PMID: 15164767 DOI: 10.1016/j.bbalip.2003.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Revised: 10/10/2003] [Accepted: 10/23/2003] [Indexed: 11/19/2022]
Abstract
Based on the assumption that fatty-acid-binding proteins (FABPs) of the epidermal-type (E-FABP) and heart-type (H-FABP) in murine alveolar type II (TII) cells mediate the synthesis of dipalmitoyl phosphatidylcholine (DPPC), the main surfactant phospholipid, we analysed TII cells isolated from wild-type (wt) and E/H-FABP double-knockout (double-ko) mice. Application of labelled palmitic acid to these cells revealed a drop in uptake, beta-oxidation, and incorporation into neutral lipids and total phosphatidylcholine (PC) of TII cells from double-ko mice. Whereas incorporation of labelled palmitic acid into DPPC remained unchanged, degradation studies demonstrated a substantial shift in DPPC synthesis from de novo to reacylation. In addition, increased expression of mRNAs and proteins of caveolin-1 and PPARgamma, and an increase of the mRNA encoding fatty acid translocase (FAT) was observed in the double-ko phenotype. As caveolin-1 interacted with PPARgamma, we assumed that FAT, caveolin-1, and PPARgamma form a signalling chain for fatty acid or drug. Consequently, PPARgamma-selective pioglitazone was added to the diet of double-ko mice. We found that further activation of PPARgamma could 'heal' the E/H-FABP double-ko effect in these TII cells as transport and utilisation of labelled palmitic acid restored a wt phenocopy. This indicated that E-FABP and/or H-FABP are involved in the mediation of DPPC synthesis in wt TII cells.
Collapse
Affiliation(s)
- Florian Guthmann
- Clinic for Neonatology, Charité Campus Mitte, Humboldt-Universität zu Berlin, 10098 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Voldstedlund M, Thuneberg L, Tranum-Jensen J, Vinten J, Christensen EI. Caveolae, caveolin and cav-p60 in smooth muscle and renin-producing cells in the rat kidney. ACTA ACUST UNITED AC 2003; 179:179-88. [PMID: 14510782 DOI: 10.1046/j.1365-201x.2003.01183.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIMS In vascular smooth muscle cells caveolae are important for signalling mechanisms regulating vascular contraction. In smooth muscle layer of the renal afferent arteriole juxtaglomerular cells (JG cells) are non-contractile renin producing cells that have the capacity to change their phenotype into smooth muscle cells and back again by metaplastic transformation. Signalling mechanisms in JG cells are not fully understood and we therefore investigated if caveolae were present, and thereby could be involved as integrators of cellular signalling in both of these phenotypes of smooth muscle cells. METHODS Using electron microscopy we compared the number of caveolae in JG cells and smooth muscle cells in the afferent arteriole of the rat kidney. The expression of caveolin and cav-p60 was examined using a combination of immunogold electron microscopy and immunofluorescence microscopy. RESULTS We found that JG cells have sixfold less caveolae per cell surface sectional length than smooth muscle cells. The expression of cavolin-1 and cav-p60 correlated with the number of caveolae. An examination of the general distribution of caveolae, cav-p60 and caveolins in the rat kidney showed that cav-p60, like caveolin-1, is a specific maker of caveolae. CONCLUSION The number of caveolae in JG cells is very low, and this makes it unlikely that caveolae are of major importance for the renin secretion specific for JG cells.
Collapse
MESH Headings
- Animals
- Blotting, Western/methods
- Caveolae/metabolism
- Caveolins/analysis
- Immunohistochemistry/methods
- Kidney/cytology
- Kidney/metabolism
- Kidney/ultrastructure
- Male
- Microscopy, Electron/methods
- Microscopy, Fluorescence/methods
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Rats
- Rats, Wistar
- Renin/biosynthesis
Collapse
Affiliation(s)
- M Voldstedlund
- Department of Medical Physiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
23
|
Abstract
Caveolae exist at cell surfaces as caveolin-coated invaginations that perform transport and signalling functions influencing cell growth, apoptosis, angiogenesis and transvascular exchange. Caveolin could constitute a key switch in tumour development through its function as a tumour suppressor and as a promoter of metastasis, chemoresistance and survival. Targeting of drugs and gene vectors to tissue-specific proteins in caveolae allows selective delivery into vascular endothelial cells in vivo and might even improve direct access to solid-tumour cells. Therefore, caveolae seem to be rich in potential targets for cancer imaging and therapeutics.
Collapse
Affiliation(s)
- Lucy A Carver
- Sidney Kimmel Cancer Center, Division of Vascular Biology and Angiogenesis, 10835 Altman Row, San Diego, California 92121, USA
| | | |
Collapse
|
24
|
Chao WT, Fan SS, Chen JK, Yang VC. Visualizing caveolin-1 and HDL in cholesterol-loaded aortic endothelial cells. J Lipid Res 2003; 44:1094-9. [PMID: 12639973 DOI: 10.1194/jlr.m300033-jlr200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Caveolae are vesicular invaginations of the plasma membranes that regulate signal transduction and transcytosis, as well as cellular cholesterol homeostasis. Our previous studies indicated that the removal of cholesterol from aortic endothelial cells and smooth muscle cells in the presence of HDL is associated with plasmalemmal invaginations and plasmalemmal vesicles. The goal of the present study was to investigate the location and distribution of caveolin-1, the main structural protein component of caveolae, in cholesterol-loaded aortic endothelial cells after HDL incubation. Confocal microscopic analysis demonstrated that the caveolin-1 appeared to colocalize with HDL-fluorescein 1,1'-dioctadecyl 3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) conjugates on the cell surface. No free HDL-DiI conjugates were revealed in the cytoplasm. Immunoelectron microscopy further demonstrated that caveolin-1 gold (15 nm) conjugates colocalized with HDL gold (10 nm) conjugates in the plasmalemmal invaginations. These morphological results indicated that caveolae are the major membrane domains facilitating the transport of excess cholesterol to HDL on the cell surface of aortic endothelial cells.
Collapse
Affiliation(s)
- W T Chao
- Department of Biology and Life Science Research Center, Tunghai University, Taichung, Taiwan, Republic of China
| | | | | | | |
Collapse
|
25
|
Abstract
Almost 50 years after the first sighting of small pits that covered the surface of mammalian cells, investigators are now getting to grips with the detailed workings of these enigmatic structures that we now know as caveolae.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience and Centre for Functional and Applied Genomics, Centre for Microscopy and Microanalysis, and School of Biomedical Sciences, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
26
|
Feng D, Nagy JA, Dvorak HF, Dvorak AM. Ultrastructural studies define soluble macromolecular, particulate, and cellular transendothelial cell pathways in venules, lymphatic vessels, and tumor-associated microvessels in man and animals. Microsc Res Tech 2002; 57:289-326. [PMID: 12112440 DOI: 10.1002/jemt.10087] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We present de novo studies and review published efforts from our laboratory, spanning 12 years (from 1988 to 2000), where we have used ultrastructural approaches to study the functional anatomy of the microvasculature in man and animals in health and disease. These efforts have defined a new endothelial cell organelle, termed the vesiculo-vacuolar organelle (VVO), which participates in the regulated transendothelial cell passage of soluble macromolecules. The studies defining this organelle utilized ultrathin serial sections, three-dimensional computer-assisted reconstructions, and ultrastructural electron-dense tracers to establish luminal to abluminal transendothelial cell continuity of VVOs. Commonality of VVOs and caveolae is suggested by the ultrastructural anatomy of individual units of VVOs and caveolae, the presence of caveolin in both structures, and a mathematical analysis of morphometric data, all of which suggest that VVOs form from fusions of individual size units equivalent to vesicles of caveolar size. Ultrastructural studies have localized potent permeability factors and their specific receptors to VVOs in in vivo tumor and allergic inflammation models. Regulation of permeability through VVOs has been quantified and shown to be increased in tumor microvessels and in control vessels exposed to potent permeability-inducing mediators. The transendothelial cell passage of particulate macromolecules occurs by vacuolar transport in tumor vessels; in permeability factor-exposed control vessels, colloidal carbon traversed endothelial cells via the development of pores that did not communicate with or disrupt intercellular junctions by gap formation. Serial section and computer-assisted reconstructions established these findings and suggested the possible development of transendothelial cell pores from VVOs. Serial sectioning and computer-assisted three-dimensional reconstructions of ultrastructural samples of an acute inflammation model revealed a transendothelial cell traffic route for motile neutrophils and platelets in the absence of classical ultrastructural criteria for regulated secretion from either cell.
Collapse
Affiliation(s)
- Dian Feng
- Department of Pathology, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
27
|
Abstract
Transport of plasma soluble constituents across the capillary wall is of primordial importance in cardiovascular physiology. While physiological experiments have concluded with the existence of two sets of pores, a large one responsible for the transport of proteins and a small one designed for the diffusion of small solutes, the morphological counterparts have yet to get general agreement. In this review, we present the different proposed paths within and between the endothelial cells that do allow passage of plasma constituents and may respond to the definitions established by physiological means. The vesicular system existing in endothelial cells has been the first transendothelial path to be proposed. Several data have demonstrated the involvement of this system in transport, although others have systematically brought controversy. One alternative to the vesicles has been the demonstration of membrane-bound tubules creating, in certain cases, transendothelial channels that would allow diffusion of plasma proteins and other constituents across the capillary wall. Access to this tubulo-vesicular system could be restrained by the stomatal diaphragm and facilitated by specific membrane receptors. Further, we have demonstrated for the first time with morpho-cytochemical tools, that the intercellular clefts are the site of diffusion for small molecules such as peptides having a molecular weight inferior to 3,000. For the fenestrated capillary bed, we have shown that fenestrae are the site through which plasma constituents cross the capillary wall. However, and in spite of the existence of these large open pores, the endothelial cells still display the tubulo-vesicular system involved in transport of large molecules and their intercellular clefts are also the site of diffusion of small molecules. Making consensus on the existence of an intracellular tubulo-vesicular system in non-fenestrated capillaries, responsible for the transport of large molecules by the endothelial cells, and understanding the rational for the fenestrated capillary to have three paths for transport--the fenestrae, the tubulo-vesicular system, and the inter-endothelial clefts--require further investigation.
Collapse
Affiliation(s)
- Moise Bendayan
- Department of Pathology and Cell Biology, Université de Montreal, Montreal, Quebec, Canada H3C 3J7.
| |
Collapse
|
28
|
Abstract
Caveolae are spherical invaginations of the plasma membrane and associated vesicles that are found at high surface densities in most cells, endothelia included. Their structural framework has been shown to consist of oligomerized caveolin molecules interacting with cholesterol and sphingolipids. Caveolae have been involved in many cellular functions such as endocytosis, signal transduction, mechano-transduction, potocytosis, and cholesterol trafficking. Some confusion still persists in the field with respect to the relationship between caveolae and the lipid rafts, which have been involved in many of the above functions. In addition to all these, endothelial caveolae have been involved in capillary permeability by their participation in the process of transcytosis. This short review will focus on their structure and components, methods used to determine these components, and the role of caveolae in the transendothelial exchanges between blood plasma and the interstitial fluid.
Collapse
Affiliation(s)
- Radu-Virgil Stan
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093-0651, USA.
| |
Collapse
|
29
|
McIntosh DP, Tan XY, Oh P, Schnitzer JE. Targeting endothelium and its dynamic caveolae for tissue-specific transcytosis in vivo: a pathway to overcome cell barriers to drug and gene delivery. Proc Natl Acad Sci U S A 2002; 99:1996-2001. [PMID: 11854497 PMCID: PMC122308 DOI: 10.1073/pnas.251662398] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2001] [Accepted: 12/10/2001] [Indexed: 11/18/2022] Open
Abstract
Site-directed pharmacodelivery is a desirable but elusive goal. Endothelium and epithelium create formidable barriers to endogenous molecules as well as targeted therapies in vivo. Caveolae provide a possible, yet unproven, transcellular pathway to overcome such barriers. By using an antibody- and subfractionation-based strategy, we generated a monoclonal antibody specific for lung caveolae (TX3.833) that targets rat lungs after i.v. injection (up to 89% of dose in 30 min). Unlike control antibodies (nonbinding or to lipid rafts), TX3.833 targets lung caveolae that bud to form free vesicles for selective and quantal transendothelial transport to underlying tissue cells in vivo. Rapid sequential transcytosis can occur to the alveolar air space via epithelial caveolae. Conjugation to TX3.833 increases drug delivery to the lung up to 172-fold and achieves rapid, localized bioefficacy. We conclude that: (i) molecular heterogeneity of the endothelium and its caveolae permits vascular targeting to achieve theoretical expectations of tissue-specific delivery and bioefficacy; (ii) caveolae can mediate selective transcytosis in vivo; and (iii) targeting caveolae may provide a tissue-specific pathway for overcoming key cell barriers to many drug and gene therapies in vivo.
Collapse
Affiliation(s)
- Deirdre P McIntosh
- Department of Pathology, Harvard Medical School, Beth Israel Hospital, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
30
|
Schubert W, Frank PG, Razani B, Park DS, Chow CW, Lisanti MP. Caveolae-deficient endothelial cells show defects in the uptake and transport of albumin in vivo. J Biol Chem 2001; 276:48619-22. [PMID: 11689550 DOI: 10.1074/jbc.c100613200] [Citation(s) in RCA: 252] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of endothelial cell caveolae in the uptake and transport of macromolecules from the blood-space to the tissue-space remains controversial. To address this issue directly, we employed caveolin-1 gene knock-out mice that lack caveolin-1 protein expression and caveolae organelles. Here, we show that endothelial cell caveolae are required for the efficient uptake and transport of a known caveolar ligand, i.e. albumin, in vivo. Caveolin-1-null mice were perfused with 5-nm gold-conjugated albumin, and its uptake was followed by transmission electron microscopy. Our results indicate that gold-conjugated albumin is not endocytosed by Cav-1-deficient lung endothelial cells and remains in the blood vessel lumen; in contrast, gold-conjugated albumin was concentrated and internalized by lung endothelial cell caveolae in wild-type mice, as expected. To quantitate this defect in uptake, we next studied the endocytosis of radioiodinated albumin using aortic ring segments from wild-type and Cav-1-null mice. Interestingly, little or no uptake of radioiodinated albumin was observed in the aortic segments from Cav-1-deficient mice, whereas aortic segments from wild-type mice showed robust uptake that was time- and temperature-dependent and competed by unlabeled albumin. We conclude that endothelial cell caveolae are required for the efficient uptake and transport of albumin from the blood to the interstitium.
Collapse
Affiliation(s)
- W Schubert
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | |
Collapse
|
31
|
RIPPE BENGT, ROSENGREN BERTINGE, VENTUROLI DANIELE. The Peritoneal Microcirculation in Peritoneal Dialysis. Microcirculation 2001. [DOI: 10.1111/j.1549-8719.2001.tb00178.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Ogawa K, Imai M, Ogawa T, Tsukamoto Y, Sasaki F. Caveolar and intercellular channels provide major transport pathways of macromolecules across vascular endothelial cells. THE ANATOMICAL RECORD 2001; 264:32-42. [PMID: 11505369 DOI: 10.1002/ar.1109] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Serum macromolecules are transported through the vascular endothelial layer to the interstitium via the caveolae and interendothelial clefts, but the nature of the permeability of these structures is unknown, and the manner of caveola-vesicle transport is controversial. We have developed a method of detecting macromolecular channels using an in situ HRP perfusion into arteries previously perfused with aldehyde and random conventional sectioning for electron microscopy. Using unbiased morphometry, 4.75% of the abluminal caveolae and 15.13% of the intercellular clefts were the tracer-positive in rat aortic endothelium. In rat aortas treated with N-ethylmaleimide, all caveolae and most free vesicles in the cytoplasm except those around the Golgi area were HRP-positive in the endothelial cells; 1.48% of abluminal caveolae were structurally recognized as caveolar channels through the endothelial layer in a plane of single section. The length density of the abluminal caveolae was decreased to about 80% to the physiological control level whereas the larger invaginations were more frequently observed. Moreover 96.17% of the intercellular clefts were HRP-positive. We suggest that a flexible channel-system functions extensively as a macromolecular transport pathway in the arterial endothelium in vivo because the tracer-labeled abluminal caveolae and intercellular clefts should be opened to the luminal surfaces methodologically. We therefore propose that caveolar channels, rather than transcytosis, provide a mechanism of caveola-vesicle transport in the endothelial cells, because free vesicles involved in transcytosis were few in number.
Collapse
Affiliation(s)
- K Ogawa
- Department of Veterinary Anatomy, Graduate School of Agriculture and Biological Sciences, Osaka Prefecture University, Osaka, Japan.
| | | | | | | | | |
Collapse
|
33
|
Schnitzer JE. Caveolae: from basic trafficking mechanisms to targeting transcytosis for tissue-specific drug and gene delivery in vivo. Adv Drug Deliv Rev 2001; 49:265-80. [PMID: 11551399 DOI: 10.1016/s0169-409x(01)00141-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Continuous endothelium and epithelium create formidable barriers to endogenous molecules as well as targeted drug and gene therapies in vivo. Caveolae represent a possible vesicular trafficking pathway through cell barriers. Here we discuss recent discoveries regarding the basic function of caveolae in transport including transcellular trafficking, intracellular trafficking to distinct endosomes, and molecular mechanisms mediating their budding, docking and fusion (dynamin and SNARE machinery). New technologies to purify and map caveolae as well as generate new probes selectively targeting caveolae in vivo provide valuable tools not only for investigating caveolar endocytosis/transcytosis but also elucidating new potential applications for site-directed treatment of many diseases. Vascular targeting of the caveolar trafficking pathway may be a useful strategy for achieving tissue-specific pharmacodelivery that also overcomes key, normally restrictive cell barriers which greatly reduce the efficacy of many therapies in vivo.
Collapse
Affiliation(s)
- J E Schnitzer
- Sidney Kimmel Cancer Center, Division of Vascular Biology and Angiogenesis, 10835 Altman Row, San Diego, CA 92121, USA.
| |
Collapse
|
34
|
Gumbleton M. Caveolae as potential macromolecule trafficking compartments within alveolar epithelium. Adv Drug Deliv Rev 2001; 49:281-300. [PMID: 11551400 DOI: 10.1016/s0169-409x(01)00142-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
With inhalational delivery the alveolar epithelium appears to be the appropriate lung surface to target for the systemic delivery of macromolecules, such as therapeutic proteins. The existence of a high numerical density of smooth-coated or non-coated plasma membrane vesicles or invaginations within the alveolar epithelial type I cell has long been recognised. The putative function of these vesicles in macromolecule transport remains the focus of research in both pulmonary physiology and pharmaceutical science disciplines. These vesicles, or subpopulations thereof, have been shown to biochemically possess caveolin, a marker protein for caveolae. This review considers the morphometric and biochemical studies that have progressed the characterisation of the vesicle populations within alveolar type I epithelium. Parallel research findings from the endothelial literature have been considered to contrast the state of progress of caveolae research in alveolar epithelium. Speculation is made on a model of caveolae vesicle-mediated transport that may satisfy some of the pulmonary pharmacokinetic data that has been generated for macromolecule absorption. The putative transport function of caveolae within alveolar epithelium is reviewed with respect to in-situ tracer studies conducted within the alveolar airspace. Finally, the functional characterisation of in-vitro alveolar epithelial cell cultures is considered with respect to the role of caveolae in macromolecule transport. A potentially significant role for alveolar caveolae in mediating the alveolar airspace to blood transport of macromolecules cannot be dismissed. Considerable research is required, however, to address this issue in a quantitative manner. A better understanding of the membrane dynamics of caveolae in alveolar epithelium will help resolve the function of these vesicular compartments and may lead to the development of more specific drug targeting approaches for promoting pulmonary drug delivery.
Collapse
Affiliation(s)
- M Gumbleton
- Pharmaceutical Cell Biology, Welsh School of Pharmacy, Redwood Building, Cardiff University, Cardiff CF10 3XF, UK.
| |
Collapse
|
35
|
Dvorak AM, Feng D. The vesiculo-vacuolar organelle (VVO). A new endothelial cell permeability organelle. J Histochem Cytochem 2001; 49:419-32. [PMID: 11259444 DOI: 10.1177/002215540104900401] [Citation(s) in RCA: 194] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A newly defined endothelial cell permeability structure, termed the vesiculo-vacuolar organelle (VVO), has been identified in the microvasculature that accompanies tumors, in venules associated with allergic inflammation, and in the endothelia of normal venules. This organelle provides the major route of extravasation of macromolecules at sites of increased vascular permeability induced by vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), serotonin, and histamine in animal models. Continuity of these large sessile structures between the vascular lumen and the extracellular space has been demonstrated in kinetic studies with ultrastructural electron-dense tracers, by direct observation of tilted electron micrographs, and by ultrathin serial sections with three-dimensional computer reconstructions. Ultrastructural enzyme-affinity cytochemical and immunocytochemical studies have identified histamine and VPF/VEGF bound to VVOs in vivo in animal models in which these mediators of permeability are released from mast cells and tumor cells, respectively. The high-affinity receptor for VPF/VEGF, VEGFR-2, was localized to VVOs and their substructural components by pre-embedding ultrastructural immunonanogold and immunoperoxidase techniques. Similar methods were used to localize caveolin and vesicle-associated membrane protein (VAMP) to VVOs and caveolae, indicating a possible commonality of formation and function of VVOs to caveolae.
Collapse
Affiliation(s)
- A M Dvorak
- The Departments of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
36
|
Lee WC, Chao WT, Yang VC. Effects of high-cholesterol diet on the interendothelial clefts and the associated junctional complexes in rat aorta. Atherosclerosis 2001; 155:307-12. [PMID: 11254900 DOI: 10.1016/s0021-9150(00)00578-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The arterial endothelial intercellular cleft (AEC) and its associated junctional complex (JC) are the determinants of permeability to macromolecules. This study analyzed frequencies of AEC and JC profile types in the rat thoracic aorta at 1 and 12 months after feeding the animals with a normal or a high-cholesterol diet. Rats on either a normal diet or high-cholesterol diet for 12 months showed more of the simple 'end to end' or 'overlap' types (P < 0.01) but fewer complex 'interdigitating' type (P < 0.01) of AEC compared to the 1 month group. With regard to JC, the frequencies of gap junctions were decreased (P < 0.01) while the tight junctions and the normal junctionless complex were increased (P < 0.01) after 12 months of normal diet as compared with 1 month on the normal diet. These changes in frequencies for gap junction and tight junction were even greater for the high-cholesterol diet than for the normal diet treatment. Moreover, the incidence of open junctions was also noticeably increased after 12 months of high-cholesterol diet. These findings suggest that the proportions of the AEC and JC were highly responsive to aging whereas those of JC were more susceptible to the high-cholesterol diet treatment.
Collapse
Affiliation(s)
- W C Lee
- Department of Biology, Tunghai University, Taichung, Taiwan, ROC
| | | | | |
Collapse
|
37
|
Feng D, Flaumenhaft R, Bandeira-Melo C, Weller P, Dvorak A. Ultrastructural localization of vesicle-associated membrane protein(s) to specialized membrane structures in human pericytes, vascular smooth muscle cells, endothelial cells, neutrophils, and eosinophils. J Histochem Cytochem 2001; 49:293-304. [PMID: 11181732 DOI: 10.1177/002215540104900303] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Vesicle-associated membrane proteins (VAMPs) are important to the trafficking of vesicles between membrane-bound intracytoplasmic organelles, in the facilitation of neurosecretion, and in constitutive and regulated secretion in non-neuronal cells. We used a pre-embedding ultrastructural immunonanogold method to localize VAMPs to subcellular sites in human cells of five lineages known to have cytoplasmic vesicles that may function in vesicular transport. We found VAMPs localized to caveolae in pericytes, vascular smooth muscle cells, and endothelial cells of venules, to the vesiculo-vacuolar organelle, recently defined in venular endothelial cells, to the vesicle-rich intergranular cytoplasm and secretory granule membranes of neutrophils, and to perigranular cytoplasmic secretory vesicles and secretory granule membranes in eosinophils. These specific localizations in five human vascular and granulocyte lineages support the notion that VAMPs have vesicle-associated functions in these cells.
Collapse
Affiliation(s)
- D Feng
- Departments of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
38
|
Abstract
This study was undertaken to evaluate the role of transcytosis as a bulk transfer mechanism for the passage of albumin from blood to tissue. Isolated rat lungs were continuously weighed and perfused with an albumin-serum buffer solution under strictly controlled hemodynamic conditions, which allowed measurements of microvascular pressure and of the capillary filtration coefficient (L(p)S). With the use of a tissue uptake technique, it was possible to determine lung albumin clearance under isogravimetric conditions (Cl(iso)), or at elevated filtration rates, to obtain an "apparent albumin reflection coefficient" (sigma(alb)). Experiments were performed during control and after reducing lung temperature from 35 degrees to 22 degrees C and after infusions of the transcytosis inhibitors N-ethylmaleimide (NEM) or filipin. Cooling moderately increased vascular resistance and reduced L(p)S and Cl(iso) largely in proportion to the induced increases in viscosity. At 35 degrees C, NEM (0.13 mM) caused a marked increase in L(p)5 and in Cl(150) and also caused a reduction in sigma(alb.) Furthermore, Cl(iso) increased for the highest dose of filipin tested (1.8 microg/ml). The demonstrated relative cooling insensitivity of the transfer of albumin across the endothelium in rat lungs does not support the contention of transcytosis of proteins across the endothelium. Furthermore, neither NEM nor filipin inhibited lung microvascular albumin transport, but actually increased lung endothelial permeability.
Collapse
Affiliation(s)
- B Rippe
- Department of Nephrology, University Hospital of Lund, S-221 85 Lund, Sweden.
| | | |
Collapse
|
39
|
Abstract
1. Macromolecules cross capillary walls via large vascular pores that are thought to be formed by plasmalemmal vesicles. Early hypotheses suggested that vesicles transferred plasma constituents across the endothelial wall either by a "shuttle" mechanism or by fusing to form transient patent channels for diffusion. Recent evidence shows that the transcytotic pathway involves both movement of vesicles within the cell and a series of fusions and fissions of the vesicular and cellular membranes. 2. The transfer of macromolecules across the capillary wall is highly specific and is mediated by receptors incorporated into specific membrane domains. Therefore, despite their morphological similarity, endothelial vesicles from heterogeneous populations in which the predominant receptor proteins incorporated in their membranes define the functions of individual vesicles. 3. Blood-brain barrier capillaries have very low permeabilities to most hydrophilic molecules. Their low permeability to macromolecules has been presumed to be due to an inhibition of the transcytotic mechanism, resulting in a low density of endothelial vesicles. 4. A comparison of vesicular densities and protein permeabilities in a number of vascular beds shows only a very weak correlation, therefore vesicle numbers alone cannot be used to predict permeability to macromolecules. 5. Blood-brain barrier capillaries are fully capable of transcytosing specific proteins, for example, insulin and transferrin, although the details are still somewhat controversial. 6. It has recently been shown that the albumin binding protein gp60 (also known as albondin), which facilitates the transcytosis of native albumin in other vascular beds, is virtually absent in brain capillaries. 7. It seems likely that the low blood-brain barrier permeability to macromolecules may be due to a low level of expression of specific receptors, rather than to an inhibition of the transcytosis mechanism.
Collapse
Affiliation(s)
- P A Stewart
- Department of Surgery, University of Toronto, Ontario, Canada.
| |
Collapse
|
40
|
Abstract
This review addresses classical questions concerning microvascular permeabiltiy in the light of recent experimental work on intact microvascular beds, single perfused microvessels, and endothelial cell cultures. Analyses, based on ultrastructural data from serial sections of the clefts between the endothelial cells of microvessels with continuous walls, conform to the hypothesis that different permeabilities to water and small hydrophilic solutes in microvessels of different tissues can be accounted for by tortuous three-dimensional pathways that pass through breaks in the junctional strands. A fiber matrix ultrafilter at the luminal entrance to the clefts is essential if microvascular walls are to retain their low permeability to macromolecules. Quantitative estimates of exchange through the channels in the endothelial cell membranes suggest that these contribute little to the permeability of most but not all microvessels. The arguments against the convective transport of macromolecules through porous pathways and for the passage of macromolecules by transcytosis via mechanisms linked to the integrity of endothelial vesicles are evaluated. Finally, intracellular signaling mechanisms implicated in transient increases in venular microvessel permeability such as occur in acute inflammation are reviewed in relation to studies of the molecular mechanisms involved in signal transduction in cultured endothelial cells.
Collapse
Affiliation(s)
- C C Michel
- Cellular and Integrative Biology, Division of Biomedical Sciences, Imperial College School of Medicine, London, United Kingdom
| | | |
Collapse
|
41
|
Sörensson J, Matejka GL, Ohlson M, Haraldsson B. Human endothelial cells produce orosomucoid, an important component of the capillary barrier. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:H530-4. [PMID: 9950854 DOI: 10.1152/ajpheart.1999.276.2.h530] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The serum protein orosomucoid (alpha1-acid glycoprotein) is needed to maintain the high capillary permselectivity required for normal homeostasis. It is not known how the protein executes its action, but it seems to contribute to the charge barrier. Moreover, recent studies suggest that the endothelial glycocalyx is essential for the charge barrier. The main site of orosomucoid synthesis is the liver, but we wanted to explore the possibility that orosomucoid was synthesized in endothelial cells. Primary cultures of human microvascular endothelial cells (HMVEC) from dermal tissue were established. Human liver cells were used as positive controls, and total RNA was prepared from both cell types. Reverse transcription-polymerase chain reaction (RT-PCR) was performed and demonstrated orosomucoid expression. After RT-PCR, the identities of the PCR products were confirmed by sequencing. RNase protection assay performed on total RNA from the HMVEC confirmed the results from the RT-PCR, i.e., orosomucoid mRNA is expressed by endothelial cells. Synthesis of orosomucoid in both liver and endothelial cells was demonstrated by immunoprecipitation. In conclusion, endothelial cells normally produce orosomucoid, which is essential for capillary charge selectivity. We suggest that orosomucoid exerts its effect by interacting with other components of the endothelial glycocalyx.
Collapse
Affiliation(s)
- J Sörensson
- Department of Physiology, Göteborg University, SE-405 30 Göteborg, Sweden
| | | | | | | |
Collapse
|
42
|
Bar RS, Erondu NE. Receptor‐Mediated Endocytosis of Polypeptide Hormones by Vascular Endothelium. Compr Physiol 1998. [DOI: 10.1002/cphy.cp070103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Devuyst O, Nielsen S, Cosyns JP, Smith BL, Agre P, Squifflet JP, Pouthier D, Goffin E. Aquaporin-1 and endothelial nitric oxide synthase expression in capillary endothelia of human peritoneum. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:H234-42. [PMID: 9688919 DOI: 10.1152/ajpheart.1998.275.1.h234] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Water transport during peritoneal dialysis (PD) requires ultrasmall pores in the capillary endothelium of the peritoneum and is impaired in the case of peritoneal inflammation. The water channel aquaporin (AQP)-1 has been proposed to be the ultrasmall pore in animal models. To substantiate the role of AQP-1 in the human peritoneum, we investigated the expression of AQP-1, AQP-2, and endothelial nitric oxide synthase (eNOS) in 19 peritoneal samples from normal subjects (n = 5), uremic patients treated by hemodialysis (n = 7) or PD (n = 4), and nonuremic patients (n = 3), using Western blotting and immunostaining. AQP-1 is very specifically located in capillary and venule endothelium but not in small-size arteries. In contrast, eNOS is located in all types of endothelia. Immunoblot for AQP-1 in human peritoneum reveals a 28-kDa band (unglycosylated AQP-1) and diffuse bands of 35-50 kDa (glycosylated AQP-1). Although AQP-1 expression is remarkably stable in all samples whatever their origin, eNOS (135 kDa) is upregulated in the three patients with ascites and/or peritonitis (1 PD and 2 nonuremic patients). AQP-2, regulated by vasopressin, is not expressed at the protein level in human peritoneum. This study 1) supports AQP-1 as the molecular counterpart of the ultrasmall pore in the human peritoneum and 2) demonstrates that AQP-1 and eNOS are regulated independently of each other in clinical conditions characterized by peritoneal inflammation.
Collapse
Affiliation(s)
- O Devuyst
- Department of Nephrology, University of Louvain Medical School, B-1200 Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Wapnir RA, Wingertzahn MA, Moyse J, Teichberg S. Proabsorptive effects of modified tapioca starch as an additive of oral rehydration solutions. J Pediatr Gastroenterol Nutr 1998; 27:17-22. [PMID: 9669721 DOI: 10.1097/00005176-199807000-00004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Partially hydrolyzed starches from staple cereals, obtained by heat or by enzymatic treatment, are often used in the formulation of homemade or extemporaneously used oral rehydration solutions used in developing countries. Conflicting or anecdotal results obtained thus far could be clarified with a standardized preparation tested under well-controlled laboratory conditions. METHODS A modified commercial tapioca starch was tested. Textra (National Starch and Chemical Co. Bridgewater, NJ, U.S.A.) added at 0, 5 or 10 g/l to an oral rehydration solution with 90 mM sodium and 111 mM glucose, in 30 rats malnourished by a protein-deficient diet for 3 weeks and in 26 well-fed control animals, using a one-pass jejunal perfusion. RESULTS In protein-deficient rats, Textra stimulated sodium absorption at 5 and 10 g/l (mean +/- SEM); 0 g/l Textra: 160 +/- 13 nmol/min x cm; 5 g/l Textra: 406 +/- 31 (p < 0.0001); and 10 g/l Textra 230 +/- 27 (p < 0.02). Potassium absorption was comparably increased. Textra also improved net water absorption and the water influx:efflux ratio. Glucose absorption was increased only at 10 g/l Textra. In control rats, Textra improved sodium and net water absorption at 5 g/l, but not at 10 g/l Textra; but the influx:efflux ratio and potassium absorption were unaltered. CONCLUSIONS These data, obtained in normal and protein-deficient rats, support the view that modified starch is a potentially useful, energy-rich additive for oral rehydration solution, which does not introduce an osmotic penalty.
Collapse
Affiliation(s)
- R A Wapnir
- Department of Pediatrics and Laboratories, North Shore University Hospital-New York University School of Medicine, Manhasset 11030, USA
| | | | | | | |
Collapse
|
45
|
Abstract
The dynamins comprise an expanding family of ubiquitously expressed 100-kD GTPases that have been implicated in severing clathrin-coated pits during receptor-mediated endocytosis. Currently, it is unclear whether the different dynamin isoforms perform redundant functions or participate in distinct endocytic processes. To define the function of dynamin II in mammalian epithelial cells, we have generated and characterized peptide-specific antibodies to domains that either are unique to this isoform or conserved within the dynamin family. When microinjected into cultured hepatocytes these affinity-purified antibodies inhibited clathrin-mediated endocytosis and induced the formation of long plasmalemmal invaginations with attached clathrin-coated pits. In addition, clusters of distinct, nonclathrin-coated, flask-shaped invaginations resembling caveolae accumulated at the plasma membrane of antibody-injected cells. In support of this, caveola-mediated endocytosis of labeled cholera toxin B was inhibited in antibody-injected hepatocytes. Using immunoisolation techniques an anti-dynamin antibody isolated caveolar membranes directly from a hepatocyte postnuclear membrane fraction. Finally, double label immunofluorescence microscopy revealed a striking colocalization between dynamin and the caveolar coat protein caveolin. Thus, functional in vivo studies as well as ultrastructural and biochemical analyses indicate that dynamin mediates both clathrin-dependent endocytosis and the internalization of caveolae in mammalian cells.
Collapse
Affiliation(s)
- J R Henley
- Department of Molecular Neuroscience, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
46
|
Bendayan M, Rasio EA. Evidence of a tubular system for transendothelial transport in arterial capillaries of the rete mirabile. J Histochem Cytochem 1997; 45:1365-78. [PMID: 9313798 DOI: 10.1177/002215549704501005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The arterial endothelial cells of the rete capillaries of the eel were examined by transmission electron microscopy on thin sections, on freeze-fracture replicas, by scanning electron microscopy, after cytochemical osmium impregnation and perfusion with peroxidase. The study revealed the existence of membrane-bound tubules and vesicles that open at both the luminal and abluminal poles of the cell and at the level of the intercellular space. The tubules are straight or present successive dilations and constrictions. They branch in various directions and intrude deeply into the cell cytoplasm, forming a complex tubular network within the cell. Immunocytochemical techniques were applied on immersion-fixed tissues and on perfusion of the capillaries with albumin and insulin. These demonstrated that the tubular-vesicular system is involved in the transport of circulating proteins. Furthermore, protein A-gold immunocytochemistry has revealed the association of actin with the membranes of this system. On the basis of these results, we suggest that the transendothelial transport of serum proteins takes place by a transcytotic process through a membrane-bound tubular-vesicular system and is equivalent to the large pore system presumed from functional studies.
Collapse
Affiliation(s)
- M Bendayan
- Department of Anatomy, Université de Montréal, Quebec, Canada
| | | |
Collapse
|
47
|
Schnitzer JE, Oh P, McIntosh DP. Role of GTP hydrolysis in fission of caveolae directly from plasma membranes. Science 1996; 274:239-42. [PMID: 8824187 DOI: 10.1126/science.274.5285.239] [Citation(s) in RCA: 210] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Caveolae are specialized invaginated cell surface microdomains of undefined function. A cell-free system that reconstituted fission of caveolae from lung endothelial plasma membranes was developed. Addition of cytosol and the hydrolysis of guanosine triphosphate (GTP) induced caveolar fission. The budded caveolae were isolated as vesicles rich in caveolin and the sialoglycolipid GM1 but not glycosyl-phosphatidylinositol (GPI)-anchored proteins. These vesicles contained the molecular machinery for endocytosis and transcytosis. In permeabilized endothelial cells, GTP stimulated, whereas GTPgammaS prevented, caveolar budding and endocytosis of the cholera toxin B chain to endosomes. Thus, caveolae may bud to form discrete carrier vesicles that participate in membrane trafficking.
Collapse
Affiliation(s)
- J E Schnitzer
- Department of Pathology, Harvard Medical School, Beth Israel Hospital, Boston, MA 02215, USA.
| | | | | |
Collapse
|
48
|
Davisson RL, Bates JN, Johnson AK, Lewis SJ. Use-dependent loss of acetylcholine- and bradykinin-mediated vasodilation after nitric oxide synthase inhibition. Evidence for preformed stores of nitric oxide-containing factors in vascular endothelial cells. Hypertension 1996; 28:354-60. [PMID: 8794816 DOI: 10.1161/01.hyp.28.3.354] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the present study, we examined the possibility that the endothelium-dependent vasodilators acetylcholine and bradykinin release preformed pools of nitric oxide-containing factors. Successive injections of selected doses of acetylcholine (1.18 +/- 0.3 micrograms/kg IV) or bradykinin (5 micrograms/kg IV) caused reproducible hypotensive and vasodilator responses within sympathetically intact and sympathetically denervated hindlimbs of conscious rats. After administration of the nitric oxide synthesis inhibitor N omega-nitro-L-arginine methyl ester (L-NAME, 25 mumol/kg IV), the first injection of acetylcholine or bradykinin produced pronounced depressor and vasodilator responses that, in the case of bradykinin, were greater than those observed before L-NAME administration. However, each successive injection of acetylcholine and bradykinin produced progressively smaller responses, such that the later injections elicited a markedly diminished hypotension and vasodilation. This "use-dependent" loss of endothelium-dependent vasodilation was not due to the diminished vasorelaxant potency of nitric oxide-containing factors because the vasodilator effects of the nitric oxide donor sodium nitroprusside (32 micrograms/kg IV) and the S-nitrosothiol compound S-nitro-socysteine (200 nmol/kg IV) were augmented in the presence of L-NAME. These results suggest that the use-dependent loss of the hemodynamic effects of acetylcholine and bradykinin in L-NAME-treated rats may be due to the release and subsequent depletion of a factor whose synthesis depends on the bioavailability of nitric oxide. Taken together, these results suggest that preformed pools of nitric oxide-containing factors exist within the endothelium of resistance vessels and that endothelium-dependent agonists exert their vasorelaxant effects at least in part by the mobilization of these performed pools.
Collapse
Affiliation(s)
- R L Davisson
- Cardiovascular Center, University of Iowa, Iowa City 52242, USA
| | | | | | | |
Collapse
|
49
|
Lane NJ, Revest PA, Whytock S, Abbott NJ. Fine-structural investigation of rat brain microvascular endothelial cells: tight junctions and vesicular structures in freshly isolated and cultured preparations. JOURNAL OF NEUROCYTOLOGY 1995; 24:347-60. [PMID: 7650540 DOI: 10.1007/bf01189062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A comparison was made between endothelial cells in freshly-isolated rat brain microvessels, and following culture of the cells for 1-10 days during growth to confluence. Attention focused on tight junctions and vesicular structures, as seen in thin sections and freeze-fracture replicas. Freshly-isolated vessels had an abnormal appearance, with a profusion of luminal microvillar processes, and extensive cytoplasmic vacuolation. There were numerous vesicular profiles, reaching a density of approximately 60 microns-2, and with a large proportion open to the surface, as shown by labelling with cationized ferritin at 4 degrees C for 5 min. Junctional zones were relatively loosely organized, with evidence for some cell:cell separation, as well as some residual tight junctional sites within zonula adhaerens junctions. In freeze-fracture replicas, junctional strands showed segments of tightly packed intramembrane particles, generally on the P face. After 1 day in culture, the cells appeared more normal, with no vacuolation or luminal processes. Vesicles were still numerous, some associated with junctional zones, while tight junctions were relatively sparse; freeze-fracture showed some incomplete tight junctional strands, with some of the intramembrane particles fracturing onto the E face. The double offset fibrillar nature of the strands could occasionally be seen. Cells cultured for 4 and 10 days showed a progressive increase in the completeness of the junctional zone, with more tight junctional contacts within the length of the adhaerens junction, and an aggregation of microfilaments in the underlying cytoplasm. The number of vesicular profiles declined, and they were progressively excluded from the junctional zone. These observations have relevance for studies on the physiology of the brain endothelium in vitro, and for comparisons with the in vivo condition.
Collapse
Affiliation(s)
- N J Lane
- Physiology Department, Queen Mary & Westfield College, London, UK
| | | | | | | |
Collapse
|
50
|
Abstract
This paper describes a quantitative approach to evaluating the ultrastructural features of brain capillaries that relate to the low non-specific permeability of the blood-brain barrier (BBB). Critical features in this approach include examination of large numbers of tissue samples and consistent, objective means of measuring features of interest. Junctional clefts, i.e., continuous channels between tight junctional regions correlate well with the know vascular permeability, being low in normal adult blood-brain barrier, high in fetal brain, and high in tumours, both human and rat. Endothelial vesicles do not always correlate with vascular permeability. They have a low density in normal adult BBB, but are also low in fetal BBB and low in some intracranial tumour vessels. However, they have a high density in muscle capillaries, and others have shown that they increase in BBB vessels damaged by hypertension. Fenestrations are consistently high in leaky vessels, but not all leaky vessels have fenestrations. The density of mitochondria in endothelial cells is high in BBB vessels of some species but not in others. Glut-1, the glucose transporter of the BBB is asymmetrically distributed between the luminal and abluminal membranes of BBB capillaries, being almost four times as numerous on the abluminal face. A large intracellular pool of glucose transporter may provide a means for rapid upregulation of the surface transporters.
Collapse
Affiliation(s)
- P A Stewart
- Department of Anatomy and Cell Biology, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|